1
|
Parres-Gold J, Levine M, Emert B, Stuart A, Elowitz MB. Contextual computation by competitive protein dimerization networks. Cell 2025; 188:1984-2002.e17. [PMID: 39978343 PMCID: PMC11973712 DOI: 10.1016/j.cell.2025.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 12/03/2024] [Accepted: 01/27/2025] [Indexed: 02/22/2025]
Abstract
Many biological signaling pathways employ proteins that competitively dimerize in diverse combinations. These dimerization networks can perform biochemical computations in which the concentrations of monomer inputs determine the concentrations of dimer outputs. Despite their prevalence, little is known about the range of input-output computations that dimerization networks can perform and how it depends on network size and connectivity. Using a systematic computational approach, we demonstrate that even small dimerization networks of 3-6 monomers are expressive, performing diverse multi-input computations. Further, dimerization networks are versatile, performing different computations when their protein components are expressed at different levels, such as in different cell types. Remarkably, individual networks with random interaction affinities, when large enough, can perform nearly all potential one-input network computations merely by tuning their monomer expression levels. Thus, even the simple process of competitive dimerization provides a powerful architecture for multi-input, cell-type-specific signal processing.
Collapse
Affiliation(s)
- Jacob Parres-Gold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Matthew Levine
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Emert
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Andrew Stuart
- Division of Engineering and Applied Sciences, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
2
|
Srivastava TP, Dhar R, Karmakar S. Looking beyond the ER, PR, and HER2: what's new in the ARsenal for combating breast cancer? Reprod Biol Endocrinol 2025; 23:9. [PMID: 39833837 PMCID: PMC11744844 DOI: 10.1186/s12958-024-01338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BrCa) is a complex and heterogeneous disease with diverse molecular subtypes, leading to varied clinical outcomes and posing significant treatment challenges. The increasing global burden of BrCa, particularly in low- and middle-income countries, underscores the urgent need for more effective therapeutic strategies. The androgen receptor (AR), expressed in a substantial proportion of breast cancer cases, has emerged as a potential biomarker and therapeutic target. In breast cancer, AR exhibits diverse functions across subtypes, often interacting with other hormone receptors, thereby influencing tumor progression and treatment responses. This intricate interplay is further complicated by the presence of constitutively expressed AR splice variants (AR-Vs) that drive resistance to AR-targeting therapies through structural rearrangements in the domains and activation of aberrant signaling pathways. Although AR-targeting drugs, initially developed for prostate cancer (PCa), have shown promise in AR-positive breast cancer, significant gaps remain in understanding AR's precise functions and therapeutic potential. The systemic management of breast cancer is guided primarily by theranostic biomarkers; ER, PR, HER2, and Ki67 which also dictate the breast cancer classification. The ubiquitous expression of AR in BrCa and the emergence of AR-Vs can assist the management of disease complementing the standard of care. This article provides a comprehensive overview of AR and its splice variants in the context of breast cancer, highlighting their prognostic and predictive value across different subtypes looking beyond the conventional ER, PR, and HER2 status. This review also raises the possibility of using AR splice variants in predicting tumor aggressiveness. From the settings of developing nations, this may provide useful insight by integrating recent advances in AR-targeted therapies and exploring their translational potential, emphasizing the critical need for further research to optimize AR-based therapeutic strategies for breast cancer management.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/therapy
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Female
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
Collapse
Affiliation(s)
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
3
|
Steinkamp R, Tsitsiridis G, Brauner B, Montrone C, Fobo G, Frishman G, Avram S, Oprea T, Ruepp A. CORUM in 2024: protein complexes as drug targets. Nucleic Acids Res 2025; 53:D651-D657. [PMID: 39526397 PMCID: PMC11701639 DOI: 10.1093/nar/gkae1033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
CORUM (https://mips.helmholtz-muenchen.de/corum/) is a public database that offers comprehensive information about mammalian protein complexes, including their subunits, functions and associations with human diseases. The newly released CORUM 5.0, encompassing 7193 protein complexes, is the largest dataset of manually curated mammalian protein complexes publicly available. This update represents the most significant upgrade to the database in >15 years. At present, the molecular processes in cells that are influenced by drugs are only incompletely understood. In this latest release, we have begun systematically investigating the impact of drugs on protein complexes. Our studies are based on a dataset from DrugCentral comprising 725 protein drug targets with approved drugs and known mechanisms of action. To date, we have identified 1975 instances from the literature where a drug affects the formation and/or function of a protein complex. Numerous examples highlight the crucial role of understanding drug-protein complex relationships in drug efficacy. The expanded dataset and the inclusion of drug effects on protein complexes are expected to significantly enhance the utility and application potential of CORUM 5.0 in fields such as network medicine and pharmacological research.
Collapse
Affiliation(s)
- Ralph Steinkamp
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - George Tsitsiridis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - Barbara Brauner
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - Corinna Montrone
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - Gisela Fobo
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - Goar Frishman
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| | - Sorin Avram
- Department of Computational Chemistry, “Coriolan Dragulescu” Institute of Chemistry, 24 Mihai Viteazu Blvd, Timisoara, Timis 300223, Romania
| | - Tudor I Oprea
- Expert Systems Inc., 12730 High Bluff Drive, San Diego, CA 92130, USA
| | - Andreas Ruepp
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, Neuherberg D-85764, Germany
| |
Collapse
|
4
|
Agnoletto A, Brisken C. Hormone Signaling in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:279-307. [PMID: 39821031 DOI: 10.1007/978-3-031-70875-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Hormones control normal breast development and function. They also impinge on breast cancer (BC) development and disease progression in direct and indirect ways. The major ovarian hormones, estrogens and progesterone, have long been established as key regulators of mammary gland development in rodents and linked to human disease. However, their roles have been difficult to disentangle because they act on multiple tissues and can act directly and indirectly on different cell types in the breast, and their receptors interact at different levels within the target cell. Estrogens are well-recognized drivers of estrogen receptor-positive (ER+) breast cancers, and the ER is successfully targeted in ER+ disease. The role of progesterone receptor (PR) as a potential target to be activated or inhibited is debated, and androgen receptor (AR) signaling has emerged as a potentially interesting pathway to target on the stage.In this chapter, we discuss hormone signaling in normal breast development and in cancer, with a specific focus on the key sex hormones: estrogen, progesterone, and testosterone. We will highlight the complexities of endocrine control mechanisms at the organismal, tissue, cellular, and molecular levels. As we delve into the mechanisms of action of hormone receptors, their interplay and their context-dependent roles in breast cancer will be discussed. Drawing insights from new preclinical models, we will describe the lessons learned and the current challenges in understanding hormone action in breast cancer.
Collapse
Affiliation(s)
- Andrea Agnoletto
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Sun G, Zhao C, Han J, Wu S, Chen Y, Yao J, Li L. Regulatory mechanisms of steroid hormone receptors on gene transcription through chromatin interaction and enhancer reprogramming. Cell Oncol (Dordr) 2024; 47:2073-2090. [PMID: 39543064 DOI: 10.1007/s13402-024-01011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Regulation of steroid hormone receptors (SHRs) on transcriptional reprogramming is crucial for breast cancer progression. SHRs, including estrogen receptor (ER), androgen receptor (AR), progesterone receptor (PR), and glucocorticoid receptor (GR) play key roles in remodeling the transcriptome of breast cancer cells. However, the molecular mechanisms by which SHRs regulate chromatin landscape in enhancer regions and transcription factor interactions remain largely unknown. In this review, we summarized the regulatory effects of 3 types of SHRs (AR, PR, and GR) on gene transcription through chromatin interactions and enhancer reprogramming. Specifically, AR and PR exhibit bi-directional regulatory effects (both inhibitory and promoting) on ER-mediated gene transcription, while GR modulates the transcription of pro-proliferation genes in ER-positive breast cancer cells. In addition, we have presented four enhancer reprogramming mechanisms (transcription factor cooperation, pioneer factor binding, dynamic assisted loading, and tethering) and the multiple enhancer-promoter contact models. Based on these mechanisms and models, this review proposes that the combination of multiple therapy strategies such as agonists/antagonists of SHRs plus endocrine therapy and the adoption of the latest sequencing technologies are expected to improve the efficacy of ER positive breast cancer treatment.
Collapse
Affiliation(s)
- Ge Sun
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Chunguang Zhao
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Jing Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P.R. China
| | - Shaoya Wu
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Yan Chen
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Jing Yao
- Cancer Center, Institute of Radiation Oncology, Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| | - Li Li
- Gene Regulation and Diseases Lab, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, PR China.
| |
Collapse
|
6
|
Glaviano A, Wander SA, Baird RD, Yap KCH, Lam HY, Toi M, Carbone D, Geoerger B, Serra V, Jones RH, Ngeow J, Toska E, Stebbing J, Crasta K, Finn RS, Diana P, Vuina K, de Bruin RAM, Surana U, Bardia A, Kumar AP. Mechanisms of sensitivity and resistance to CDK4/CDK6 inhibitors in hormone receptor-positive breast cancer treatment. Drug Resist Updat 2024; 76:101103. [PMID: 38943828 DOI: 10.1016/j.drup.2024.101103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/10/2024] [Indexed: 07/01/2024]
Abstract
Cell cycle dysregulation is a hallmark of cancer that promotes eccessive cell division. Cyclin-dependent kinase 4 (CDK4) and cyclin-dependent kinase 6 (CDK6) are key molecules in the G1-to-S phase cell cycle transition and are crucial for the onset, survival, and progression of breast cancer (BC). Small-molecule CDK4/CDK6 inhibitors (CDK4/6i) block phosphorylation of tumor suppressor Rb and thus restrain susceptible BC cells in G1 phase. Three CDK4/6i are approved for the first-line treatment of patients with advanced/metastatic hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) BC in combination with endocrine therapy (ET). Though this has improved the clinical outcomes for survival of BC patients, there is no established standard next-line treatment to tackle drug resistance. Recent studies suggest that CDK4/6i can modulate other distinct effects in both BC and breast stromal compartments, which may provide new insights into aspects of their clinical activity. This review describes the biochemistry of the CDK4/6-Rb-E2F pathway in HR+ BC, then discusses how CDK4/6i can trigger other effects in BC/breast stromal compartments, and finally outlines the mechanisms of CDK4/6i resistance that have emerged in recent preclinical studies and clinical cohorts, emphasizing the impact of these findings on novel therapeutic opportunities in BC.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richard D Baird
- Cancer Research UK Cambridge Centre, Hills Road, Cambridge CB2 0QQ, UK
| | - Kenneth C-H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Masakazu Toi
- School of Medicine, Kyoto University, Kyoto, Japan
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, Inserm U1015, Université Paris-Saclay, Villejuif, France
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Robert H Jones
- Cardiff University and Velindre Cancer Centre, Museum Avenue, Cardiff CF10 3AX, UK
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine (LKCMedicine), Nanyang Technological University, Experimental Medicine Building, 636921, Singapore; Cancer Genetics Service (CGS), National Cancer Centre Singapore, 168583, Singapore
| | - Eneda Toska
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge, UK; Division of Cancer, Imperial College London, Hammersmith Campus, London, UK
| | - Karen Crasta
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore; Healthy Longetivity Translational Program, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Richard S Finn
- Department of Oncology, Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo 90123, Italy
| | - Karla Vuina
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Robertus A M de Bruin
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Uttam Surana
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; SiNOPSEE Therapeutics Pte Ltd, A⁎STARTCentral, 139955, Singapore
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
7
|
Mamczarz J, Lane M, Merchenthaler I. Letrozole delays acquisition of water maze task in female BALB/c mice: Possible involvement of anxiety. Horm Behav 2024; 162:105524. [PMID: 38513526 PMCID: PMC11155665 DOI: 10.1016/j.yhbeh.2024.105524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Letrozole, an aromatase inhibitor preventing estrogen synthesis from testosterone, is used as an adjuvant therapy in estrogen receptor-positive breast cancer patients. However, like other aromatase inhibitors, it induces many side effects, including impaired cognition. Despite its negative effect in humans, results from animal models are inconsistent and suggest that letrozole can either impair or improve cognition. Here, we studied the effects of chronic letrozole treatment on cognitive behavior of adult female BALB/c mice, a relevant animal model for breast cancer studies, to develop an appropriate animal model aimed at testing therapies to mitigate side effects of letrozole. In Morris water maze, letrozole 0.1 mg/kg impaired reference learning and memory. Interestingly, most of the letrozole 0.1 mg/kg-treated mice were able to learn the new platform position in reversal training and performed similar to control mice in a reversal probe test. Results of the reversal test suggest that letrozole did not completely disrupt spatial navigation, but rather delayed acquisition of spatial information. The delay might be related to increased anxiety as suggested by increased thigmotactic behavior during the reference memory training. The learning impairment was water maze-specific since we did not observe impairment in other spatial tasks such as in Y-maze or object location test. In contrast, the dose of 0.3 mg/kg did not have effect on water maze learning and facilitated locomotor habituation and recognition in novel object recognition test. The current study shows that letrozole dose-dependently modulates behavioral response and that its effects are task-dependent.
Collapse
Affiliation(s)
- Jacek Mamczarz
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| | - Malcolm Lane
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Istvan Merchenthaler
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| |
Collapse
|
8
|
de Nys R, van Eyk CL, Ritchie T, Møller RS, Scheffer IE, Marini C, Bhattacharjee R, Kumar R, Gecz J. Multiomic analysis implicates nuclear hormone receptor signalling in clustering epilepsy. Transl Psychiatry 2024; 14:65. [PMID: 38280856 PMCID: PMC10821879 DOI: 10.1038/s41398-024-02783-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/07/2024] [Accepted: 01/16/2024] [Indexed: 01/29/2024] Open
Abstract
Clustering Epilepsy (CE) is an epileptic disorder with neurological comorbidities caused by heterozygous variants of the X chromosome gene Protocadherin 19 (PCDH19). Recent studies have implicated dysregulation of the Nuclear Hormone Receptor (NHR) pathway in CE pathogenesis. To obtain a comprehensive overview of the impact and mechanisms of loss of PCDH19 function in CE pathogenesis, we have performed epigenomic, transcriptomic and proteomic analysis of CE relevant models. Our studies identified differential regulation and expression of Androgen Receptor (AR) and its targets in CE patient skin fibroblasts. Furthermore, our cell culture assays revealed the repression of PCDH19 expression mediated through ERα and the co-regulator FOXA1. We also identified a protein-protein interaction between PCDH19 and AR, expanding upon the intrinsic link between PCDH19 and the NHR pathway. Together, these results point to a novel mechanism of NHR signaling in the pathogenesis of CE that can be explored for potential therapeutic options.
Collapse
Affiliation(s)
- Rebekah de Nys
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Clare L van Eyk
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tarin Ritchie
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine (member of ERN EpiCARE), Danish Epilepsy Centre, Filadelfia, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Ingrid E Scheffer
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg, VIC, 3084, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Neurology, The Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Carla Marini
- Child Neurology and Psychiatry Unit Children's Hospital "G. Salesi" Azienda Ospedaliero-Universitaria delle Marche Ancona, Ancona, Italy
| | - Rudrarup Bhattacharjee
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Raman Kumar
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jozef Gecz
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, SA, 5005, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
9
|
Valente-Santos J, Vitorino R, Sousa-Mendes C, Oliveira P, Colaço B, Faustino-Rocha AI, Neuparth MJ, Leite-Moreira A, Duarte JA, Ferreira R, Amado F. Long-Term Exposure to Supraphysiological Levels of Testosterone Impacts Rat Submandibular Gland Proteome. Int J Mol Sci 2023; 25:550. [PMID: 38203721 PMCID: PMC10778877 DOI: 10.3390/ijms25010550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The salivary glands play a central role in the secretion of saliva, whose composition and volume affect oral and overall health. A lesser-explored dimension encompasses the possible changes in salivary gland proteomes in response to fluctuations in sex hormone levels. This study aimed to examine the effects of chronic exposure to testosterone on salivary gland remodeling, particularly focusing on proteomic adaptations. Therefore, male Wistar rats were implanted with subcutaneous testosterone-releasing devices at 14 weeks of age. Their submandibular glands were histologically and molecularly analyzed 47 weeks later. The results underscored a significant increase in gland mass after testosterone exposure, further supported by histologic evidence of granular duct enlargement. Despite increased circulating sex hormones, there was no detectable shift in the tissue levels of estrogen alpha and androgen receptors. GeLC-MS/MS and subsequent bioinformatics identified 308 proteins in the submandibular glands, 12 of which were modulated by testosterone. Of note was the pronounced upregulation of Klk3 and the downregulation of Klk6 and Klk7 after testosterone exposure. Protein-protein interaction analysis with the androgen receptor suggests that Klk3 is a potential target of androgenic signaling, paralleling previous findings in the prostate. This exploratory analysis sheds light on the response of salivary glands to testosterone exposure, providing proteome-level insights into the associated weight and histological changes.
Collapse
Affiliation(s)
- João Valente-Santos
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Cláudia Sousa-Mendes
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.-M.); (A.L.-M.)
| | - Paula Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (P.O.); (A.I.F.-R.)
| | - Bruno Colaço
- Veterinary and Animal Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Department of Animal Science, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Ana I. Faustino-Rocha
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal; (P.O.); (A.I.F.-R.)
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology, University of Évora, 7006-554 Évora, Portugal
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; (M.J.N.); (J.A.D.)
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
| | - Adelino Leite-Moreira
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (C.S.-M.); (A.L.-M.)
| | - José Alberto Duarte
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; (M.J.N.); (J.A.D.)
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.-S.); (R.F.)
| |
Collapse
|
10
|
Proffitt MR, Liu X, Ortlund EA, Smith GT. Evolution of androgen receptors contributes to species variation in androgenic regulation of communication signals in electric fishes. Mol Cell Endocrinol 2023; 578:112068. [PMID: 37714403 PMCID: PMC10695101 DOI: 10.1016/j.mce.2023.112068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Hormones and receptors coevolve to generate species diversity in hormone action. We compared the structure and function of androgen receptors (ARs) across fishes, with a focus on ARs in ghost knifefishes (Apteronotidae). Apteronotids, like many other teleosts, have two ARs (ARα and ARβ). ARβ is largely conserved, whereas ARα sequences vary considerably across species. The ARα ligand binding domain (LBD) has evolved under positive selection, and differences in the LBD across apteronotid species are associated with diversity in androgenic regulation of behavior. The Apteronotus leptorhynchus ARα LBD differs substantially from that of the Apteronotus albifrons ARα or the ancestral AR. Structural modeling and transactivation assays demonstrated that A. leptorhynchus ARα cannot bind androgens. We propose a model whereby relative expression of ARα versus ARβ in the brain, coupled with loss of androgen binding by ARα in A. leptorhynchus might explain reversals in androgenic regulation and sex differences in electrocommunication behavior.
Collapse
Affiliation(s)
- Melissa Renee Proffitt
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Xu Liu
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta, GA, USA
| | - G Troy Smith
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
11
|
Raj A, Chakole S, Agrawal S, Gupta A, Khekade H, Prasad R, Lohakare T, Wanjari M. The Impact of Menopause on Cardiovascular Aging: A Comprehensive Review of Androgen Influences. Cureus 2023; 15:e43569. [PMID: 37719547 PMCID: PMC10503403 DOI: 10.7759/cureus.43569] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Menopause represents a critical life stage in women, characterized by hormonal changes that significantly impact cardiovascular health. While the decline in estrogen levels has long been recognized as a major contributor to cardiovascular aging in menopausal women, the role of androgens, particularly testosterone, has gained increasing attention in recent years. This comprehensive review aims to provide a thorough understanding of the impact of menopause on cardiovascular aging, with a specific focus on the influences of androgens. A literature search was conducted to gather relevant studies and clinical evidence exploring the relationship between menopause, androgens, and cardiovascular health. The review integrates findings from various studies to present a holistic view of the topic. The review outlines the changes in hormone levels during menopause and discusses the cardiovascular risk factors associated with this transition. Furthermore, it explores the impact of menopause on cardiovascular structure and function, elucidating the underlying mechanisms that contribute to cardiovascular aging. Androgens' significance in maintaining cardiovascular homeostasis is discussed, followed by exploring the effects of androgen decline during menopause on lipid profiles, insulin sensitivity, vascular function, and other cardiovascular parameters. The review delves into the mechanisms of androgen action on the cardiovascular system, emphasizing the role of androgen receptors and the intricate interplay between androgens, estrogens, and other hormones. Clinical evidence supporting the effects of androgens on cardiovascular aging is presented, including studies investigating the association between androgen levels and cardiovascular outcomes. Additionally, the impact of androgen replacement therapy (ART) on cardiovascular risk markers and events in menopausal women is examined, along with controversies and conflicting findings surrounding the use of androgen therapy in cardiovascular aging. This structured review provides a comprehensive understanding of the impact of menopause on cardiovascular aging, with a specific focus on the role of androgens. By highlighting the significance of androgens in cardiovascular health during menopause, this review aims to create an initial impression and interest among readers, inviting potential citations in the future. The findings underscore the need for further research and offer insights into managing cardiovascular aging in menopausal women, including lifestyle interventions, pharmacological approaches, and the potential role of personalized medicine and precision therapies.
Collapse
Affiliation(s)
- Aditya Raj
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Swarupa Chakole
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Suyash Agrawal
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anannya Gupta
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Harshal Khekade
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roshan Prasad
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tejaswee Lohakare
- Pediatrics, Smt. Radhikabai Meghe Memorial College of Nursing, Wardha, IND
| | - Mayur Wanjari
- Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
12
|
Zhou FH, Downton T, Freelander A, Hurwitz J, Caldon CE, Lim E. CDK4/6 inhibitor resistance in estrogen receptor positive breast cancer, a 2023 perspective. Front Cell Dev Biol 2023; 11:1148792. [PMID: 37035239 PMCID: PMC10073728 DOI: 10.3389/fcell.2023.1148792] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
CDK4/6 inhibitors have become game-changers in the treatment of estrogen receptor-positive (ER+) breast cancer, and in combination with endocrine therapy are the standard of care first-line treatment for ER+/HER2-negative advanced breast cancer. Although CDK4/6 inhibitors prolong survival for these patients, resistance is inevitable and there is currently no clear standard next-line treatment. There is an urgent unmet need to dissect the mechanisms which drive intrinsic and acquired resistance to CDK4/6 inhibitors and endocrine therapy to guide the subsequent therapeutic decisions. We will review the insights gained from preclinical studies and clinical cohorts into the diverse mechanisms of CDK4/6 inhibitor action and resistance, and highlight potential therapeutic strategies in the context of CDK4/6 inhibitor resistance.
Collapse
Affiliation(s)
- Fiona H. Zhou
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Teesha Downton
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Allegra Freelander
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Joshua Hurwitz
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - C. Elizabeth Caldon
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| |
Collapse
|
13
|
Wei L, Gao H, Yu J, Zhang H, Nguyen TTL, Gu Y, Passow MR, Carter JM, Qin B, Boughey JC, Goetz MP, Weinshilboum RM, Ingle JN, Wang L. Pharmacological Targeting of Androgen Receptor Elicits Context-Specific Effects in Estrogen Receptor-Positive Breast Cancer. Cancer Res 2023; 83:456-470. [PMID: 36469363 PMCID: PMC9896025 DOI: 10.1158/0008-5472.can-22-1016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Androgen receptor (AR) is expressed in 80% to 90% of estrogen receptor α-positive (ER+) breast cancers. Accumulated evidence has shown that AR is a tumor suppressor and that its expression is associated with improved prognosis in ER+ breast cancer. However, both a selective AR agonist (RAD140) and an AR inhibitor (enzalutamide, ENZ) have shown a therapeutic effect on ER+ breast cancer, so the potential for clinical application of AR-targeting therapy for ER+ breast cancer is still in dispute. In this study, we evaluated the efficacy of ENZ and RAD140 in vivo and in vitro in AR+/ER+ breast cancer models, characterizing the relationship of AR and ER levels to response to AR-targeting drugs and investigating the alterations of global gene expression and chromatin binding of AR and ERα after ENZ treatment. In the AR-low setting, ENZ directly functioned as an ERα antagonist. Cell growth inhibition by ENZ in breast cancer with low AR expression was independent of AR and instead dependent on ER. In AR-high breast cancer models, AR repressed ERα signaling and ENZ promoted ERα signaling by antagonizing AR. In contrast, RAD140 activated AR signaling and suppressed AR-high tumor growth by deregulating ERα expression and blocking ERα function. Overall, analysis of the dynamic efficacies and outcomes of AR agonist, and antagonist in the presence of different AR and ERα levels reveals regulators of response and supports the clinical investigation of ENZ in selected ER+ tumors with a low AR/ER ratio and AR agonists in tumors with a high AR/ER ratio. SIGNIFICANCE The ratio of androgen receptor to estrogen receptor in breast cancer dictates the response to AR-targeted therapies, providing guidelines for developing AR-directed treatment strategies for patients with breast cancer.
Collapse
Affiliation(s)
- Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huan Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Thanh Thanh L. Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yayun Gu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Marie R. Passow
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Bo Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Matthew P. Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - James N. Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
14
|
Kolyvas EA, Caldas C, Kelly K, Ahmad SS. Androgen receptor function and targeted therapeutics across breast cancer subtypes. Breast Cancer Res 2022; 24:79. [PMID: 36376977 PMCID: PMC9664788 DOI: 10.1186/s13058-022-01574-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant progress in breast cancer (BC) therapy, it is globally the most commonly diagnosed cancer and leads to the death of over 650,000 women annually. Androgen receptor (AR) is emerging as a potential new therapeutic target in BC. While the role of AR is well established in prostate cancer (PCa), its function in BC remains incompletely understood. Emerging data show that AR's role in BC is dependent on several factors including, but not limited to, disease subtype, tumour microenvironment, and levels of circulating oestrogens and androgens. While targeting AR in PCa is becoming increasingly effective, these advances have yet to make any significant impact on the care of BC patients. However, this approach is increasingly being evaluated in BC and it is clear that improvements in our understanding of AR's role in BC will increase the likelihood of success for AR-targeted therapies. This review summarizes our current understanding of the function of AR across BC subtypes. We highlight limitations in our current knowledge and demonstrate the importance of categorizing BC subtypes effectively, in relation to determining AR activity. Further, we describe the current state of the art regarding AR-targeted approaches for BC as monotherapy or in combination with radiotherapy.
Collapse
Affiliation(s)
- Emily A Kolyvas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- NIH-Oxford-Cambridge Scholars Program, Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Breast Cancer Programme, CRUK Cambridge Centre, Cambridge, CB2 0RE, UK
- Cambridge Breast Cancer Research Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Saif S Ahmad
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK.
- Department of Oncology, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK.
| |
Collapse
|
15
|
Transcriptome profiling and proteomic validation reveals targets of the androgen receptor signaling in the BT-474 breast cancer cell line. Clin Proteomics 2022; 19:14. [PMID: 35568821 PMCID: PMC9107748 DOI: 10.1186/s12014-022-09352-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 04/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accumulating evidence suggests that the androgen receptor (AR) and its endogenous ligands influence disease progression in breast cancer (BCa). However, AR-mediated changes in BCa differ among the various BCa subtypes according to their hormone receptor profile [i.e., presence/absence of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2, (HER2)]. Thus, we explored the androgen-regulated transcriptomic changes in the ER+PR+HER2+ BCa cell line, BT-474, and compared them with PR-mediated changes. METHODS We performed RNA sequencing analysis in treated BT-474 cells with dihydrotestosterone (DHT) and progesterone. Validation of the top ten differentially androgen-regulated genes and a number of other genes found in enriched signaling pathways was performed by qRT-PCR in BT-474 and other BCa cell lines. In addition, a parallel reaction monitoring targeted proteomic approach was developed to verify selected transcripts at the protein level. RESULTS In total 19,450 transcripts were detected, of which 224 were differentially regulated after DHT treatment. The increased expression of two well-known androgen-regulated genes, KLK2 (p < 0.05) and KLK3 (p < 0.001), confirmed the successful androgen stimulation in BT-474 cells. The transcription factor, ZBTB16, was the most highly upregulated gene, with ~ 1000-fold change (p < 0.001). Pathway enrichment analysis revealed downregulation of the DNA replication processes (p < 0.05) and upregulation of the androgen signaling and fatty acid metabolism pathways (p < 0.05). Changes related to progesterone treatment showed opposite effects in gene expression than DHT treatment. Similar expression profiles were observed among other BCa cell lines expressing high levels of AR (ZR75.1 and MBA-MB-453). The parallel reaction monitoring targeted proteomic analysis further confirmed that altered protein expression (KLK3, ALOX15B) in the supernatant and cell lysate of DHT-treated BT-474 cells, compared to control cells. DISCUSSION Our findings suggest that AR modulates the metabolism of BT-474 cells by affecting the expression of a large number of genes and proteins. Based on further pathway analysis, we suggest that androgen receptor acts as a tumor suppressor in the BT-474 cells.
Collapse
|
16
|
Expression of dihydrotestosterone synthases and androgen receptor in sheep oviduct ampulla and its regulation by estradiol and progesterone. Reprod Biol 2022; 22:100573. [DOI: 10.1016/j.repbio.2021.100573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/03/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022]
|
17
|
Xia S, Lin Q. Estrogen Receptor Bio-Activities Determine Clinical Endocrine Treatment Options in Estrogen Receptor-Positive Breast Cancer. Technol Cancer Res Treat 2022; 21:15330338221090351. [PMID: 35450488 PMCID: PMC9036337 DOI: 10.1177/15330338221090351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In estrogen receptor positive (ER+) breast cancer therapy, estrogen receptors (ERs) are the major targeting molecules. ER-targeted therapy has provided clinical benefits for approximately 70% of all breast cancer patients through targeting the ERα subtype. In recent years, mechanisms underlying breast cancer occurrence and progression have been extensively studied and largely clarified. The PI3K/AKT/mTOR pathway, microRNA regulation, and other ER downstream signaling pathways are found to be the effective therapeutic targets in ER+ BC therapy. A number of the ER+ (ER+) breast cancer biomarkers have been established for diagnosis and prognosis. The ESR1 gene mutations that lead to endocrine therapy resistance in ER+ breast cancer had been identified. Mutations in the ligand-binding domain of ERα which encoded by ESR1 gene occur in most cases. The targeted drugs combined with endocrine therapy have been developed to improve the therapeutic efficacy of ER+ breast cancer, particularly the endocrine therapy resistance ER+ breast cancer. The combination therapy has been demonstrated to be superior to monotherapy in overall clinical evaluation. In this review, we focus on recent progress in studies on ERs and related clinical applications for targeted therapy and provide a perspective view for therapy of ER+ breast cancer.
Collapse
Affiliation(s)
- Song Xia
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, China
- Qiong Lin, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China.
| |
Collapse
|
18
|
Butz H, Patócs A. Mechanisms behind context-dependent role of glucocorticoids in breast cancer progression. Cancer Metastasis Rev 2022; 41:803-832. [PMID: 35761157 PMCID: PMC9758252 DOI: 10.1007/s10555-022-10047-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/09/2022] [Indexed: 02/08/2023]
Abstract
Glucocorticoids (GCs), mostly dexamethasone (dex), are routinely administered as adjuvant therapy to manage side effects in breast cancer. However, recently, it has been revealed that dex triggers different effects and correlates with opposite outcomes depending on the breast cancer molecular subtype. This has raised new concerns regarding the generalized use of GC and suggested that the context-dependent effects of GCs can be taken into potential consideration during treatment design. Based on this, attention has recently been drawn to the role of the glucocorticoid receptor (GR) in development and progression of breast cancer. Therefore, in this comprehensive review, we aimed to summarize the different mechanisms behind different context-dependent GC actions in breast cancer by applying a multilevel examination, starting from the association of variants of the GR-encoding gene to expression at the mRNA and protein level of the receptor, and its interactions with other factors influencing GC action in breast cancer. The role of GCs in chemosensitivity and chemoresistance observed during breast cancer therapy is discussed. In addition, experiences using GC targeting therapeutic options (already used and investigated in preclinical and clinical trials), such as classic GC dexamethasone, selective glucocorticoid receptor agonists and modulators, the GC antagonist mifepristone, and GR coregulators, are also summarized. Evidence presented can aid a better understanding of the biology of context-dependent GC action that can lead to further advances in the personalized therapy of breast cancer by the evaluation of GR along with the conventional estrogen receptor (ER) and progesterone receptor (PR) in the routine diagnostic procedure.
Collapse
Affiliation(s)
- Henriett Butz
- Department of Molecular Genetics and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary.
- Hereditary Tumours Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary.
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary.
| | - Attila Patócs
- Department of Molecular Genetics and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Hereditary Tumours Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Cara AL, Henson EL, Beekly BG, Elias CF. Distribution of androgen receptor mRNA in the prepubertal male and female mouse brain. J Neuroendocrinol 2021; 33:e13063. [PMID: 34866263 PMCID: PMC8711114 DOI: 10.1111/jne.13063] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
Androgens are steroid hormones that play a critical role in brain development and sexual maturation by acting upon both androgen receptors (AR) and estrogen receptors (ERα/β) after aromatization. The contribution of estrogens from aromatized androgens in brain development and the central regulation of metabolism, reproduction, and behavior is well defined, but the role of androgens acting on AR has been unappreciated. Here, we map the sex specific expression of Ar in the adult and developing mouse brain. Postnatal days (PND) 12 and 21 were used to target a critical window of prepubertal development. Consistent with previous literature in adults, sex-specific differences in Ar expression were most profound in the bed nucleus of the stria terminalis (BST), medial amygdala (MEA) and medial preoptic area (MPO). Ar expression was also high in these areas at PND 12 and 21 in both sexes. In addition, we describe extra-hypothalamic and extra-limbic areas that show moderate, consistent and similar Ar expression in both sexes at both prepubertal time points. Briefly, Ar expression was observed in olfactory areas of the cerebral cortex, the hippocampus, several thalamic nuclei, and cranial nerve nuclei involved in autonomic sensory and motor function. To further characterize forebrain populations of Ar expressing neurons and determine whether they also coexpress estrogen receptors, we examined expression of Ar, Esr1 and Esr2 in prepubertal mice in selected nuclei. We found populations of neurons in the BST, MEA and MPO that coexpress Ar, but not Esr1 or Esr2, whereas others express a combination of the three receptors. Our findings indicate that various brain areas express Ar during prepubertal development and may play an important role in female neuronal development and physiology.
Collapse
Affiliation(s)
- Alexandra L. Cara
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMIUSA
| | - Emily L. Henson
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMIUSA
| | | | - Carol F. Elias
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMIUSA
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of Obstetrics and GynaecologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
20
|
Hanamura T, Christenson JL, O'Neill KI, Rosas E, Spoelstra NS, Williams MM, Richer JK. Secreted indicators of androgen receptor activity in breast cancer pre-clinical models. Breast Cancer Res 2021; 23:102. [PMID: 34736512 PMCID: PMC8567567 DOI: 10.1186/s13058-021-01478-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Accumulating evidence has attracted attention to the androgen receptor (AR) as a biomarker and therapeutic target in breast cancer. We hypothesized that AR activity within the tumor has clinical implications and investigated whether androgen responsive serum factors might serve as a minimally invasive indicator of tumor AR activity. METHODS Based on a comprehensive gene expression analysis of an AR-positive, triple negative breast cancer patient-derived xenograft (PDX) model, 163 dihydrotestosterone (DHT)-responsive genes were defined as an androgen responsive gene set. Among them, we focused on genes that were DHT-responsive that encode secreted proteins, namely KLK3, AZGP1 and PIP, that encode the secreted factors prostate specific antigen (PSA), zinc-alpha-2-glycoprotein (ZAG) and prolactin induced protein (PIP), respectively. Using AR-positive breast cancer cell lines representing all breast cancer subtypes, expression of candidate factors was assessed in response to agonist DHT and antagonist enzalutamide. Gene set enrichment analysis (GSEA) was performed on publically available gene expression datasets from breast cancer patients to analyze the relationship between genes encoding the secreted factors and other androgen responsive gene sets in each breast cancer subtype. RESULTS Anti-androgen treatment decreased proliferation in all cell lines tested representing various tumor subtypes. Expression of the secreted factors was regulated by AR activation in the majority of breast cancer cell lines. In GSEA, the candidate genes were positively correlated with an androgen responsive gene set across breast cancer subtypes. CONCLUSION KLK3, AZGP1 and PIP are AR regulated and reflect tumor AR activity. Further investigations are needed to examine the potential efficacy of these factors as serum biomarkers.
Collapse
Affiliation(s)
- Toru Hanamura
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Emmanuel Rosas
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Michelle M Williams
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
21
|
Augusto TV, Amaral C, Almeida CF, Teixeira N, Correia-da-Silva G. Differential biological effects of aromatase inhibitors: Apoptosis, autophagy, senescence and modulation of the hormonal status in breast cancer cells. Mol Cell Endocrinol 2021; 537:111426. [PMID: 34391846 DOI: 10.1016/j.mce.2021.111426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 01/10/2023]
Abstract
Estrogen receptor-positive (ER+) breast carcinomas are the most common subtype, corresponding to 60% of the cases in premenopausal and 75% in postmenopausal women. The third-generation of aromatase inhibitors (AIs), the non-steroidal Anastrozole (Ana) and Letrozole (Let) and the steroidal Exemestane (Exe), are considered a first-line endocrine therapy for postmenopausal women. Despite their clinical success, the development of resistance is the major setback in clinical practice. Nevertheless, the lack of cross-resistance between AIs hints that these drugs may act through distinct mechanisms. Therefore, this work studied the different effects induced by AIs on biological processes, such as cell proliferation, death, autophagy and senescence. Moreover, their effects on the regulation of the hormonal environment were also explored. The non-steroidal AIs induce senescence, through increased YPEL3 expression, on aromatase-overexpressing breast cancer cells (MCF-7aro), whereas Exe promotes a cytoprotective autophagy, thus blocking senescence induction. In addition, in a hormone-enriched environment, the non-steroidal AIs prevent estrogen signaling, despite up-regulating the estrogen receptor alpha (ERα), while Exe down-regulates ERα and maintains its activation. In these conditions, all AIs up-regulate the androgen receptor (AR) which blocks EGR3 transcription in Exe-treated cells. On the other hand, in hormone-depleted conditions, a crosstalk between AR and ERα occurs, enhancing the estrogenic effects of Exe. This indicates that Exe modulates both ERα and AR, while Ana and Let act as pure AIs. Thus, this study highlights the potential clinical benefit of combining AR antagonists with Exe and discourages the sequential use of Exe as second-line therapy in postmenopausal breast cancer.
Collapse
Affiliation(s)
- Tiago V Augusto
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira N° 228, 4050-313, Porto, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira N° 228, 4050-313, Porto, Portugal
| | - Cristina F Almeida
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira N° 228, 4050-313, Porto, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira N° 228, 4050-313, Porto, Portugal.
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira N° 228, 4050-313, Porto, Portugal.
| |
Collapse
|
22
|
Kumar S, Freelander A, Lim E. Type 1 Nuclear Receptor Activity in Breast Cancer: Translating Preclinical Insights to the Clinic. Cancers (Basel) 2021; 13:4972. [PMID: 34638457 PMCID: PMC8507977 DOI: 10.3390/cancers13194972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
The nuclear receptor (NR) family of transcription factors is intimately associated with the development, progression and treatment of breast cancer. They are used diagnostically and prognostically, and crosstalk between nuclear receptor pathways and growth factor signalling has been demonstrated in all major subtypes of breast cancer. The majority of breast cancers are driven by estrogen receptor α (ER), and anti-estrogenic therapies remain the backbone of treatment, leading to clinically impactful improvements in patient outcomes. This serves as a blueprint for the development of therapies targeting other nuclear receptors. More recently, pivotal findings into modulating the progesterone (PR) and androgen receptors (AR), with accompanying mechanistic insights into NR crosstalk and interactions with other proliferative pathways, have led to clinical trials in all of the major breast cancer subtypes. A growing body of evidence now supports targeting other Type 1 nuclear receptors such as the glucocorticoid receptor (GR), as well as Type 2 NRs such as the vitamin D receptor (VDR). Here, we reviewed the existing preclinical insights into nuclear receptor activity in breast cancer, with a focus on Type 1 NRs. We also discussed the potential to translate these findings into improving patient outcomes.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| | - Allegra Freelander
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| | - Elgene Lim
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| |
Collapse
|
23
|
Jahan N, Jones C, Rahman RL. Androgen receptor expression in breast cancer: Implications on prognosis and treatment, a brief review. Mol Cell Endocrinol 2021; 531:111324. [PMID: 34000352 DOI: 10.1016/j.mce.2021.111324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023]
Abstract
Approximately 70%-85% of breast cancers express androgen receptors (ARs). The role of AR in breast cancer pathogenesis is currently in exploration. Both androgens and anti-androgens have demonstrated variable inhibitory and stimulatory effects in AR-positive breast cancer depending on estrogen receptor and HER2 co-expression. Androgen signaling pathways interact with other critical cellular pathways, such as the PI3K/AKT/mTOR, Ras/Raf/MAPK/ERK, Wnt/β-catenin, and estrogen signaling pathways. Therapeutic exploitation of AR has been the crux of management of prostate cancer for decades. In recent years there has been increasing interest in AR as a novel therapeutic target in breast cancer. There have been many early phase clinical trials evaluating the safety and efficacy of various AR-targeted agents in breast cancer. Some of these studies have shown promising clinical benefits. Studies of biomarkers to identify the patients likely to benefit from AR-targeted therapies are currently in progress. Besides, AR expression may be an important prognostic and predictive marker for breast cancer, which needs to be defined better in future studies.
Collapse
Affiliation(s)
- Nusrat Jahan
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4(th) St, Lubbock, Tx, 79430, USA.
| | - Catherine Jones
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4(th) St, Lubbock, Tx, 79430, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, Texas Tech University Health Sciences Center, 3601 4(th)St, Lubbock, Tx, 79430, USA
| |
Collapse
|
24
|
Cruz MAD, Lund D, Szekeres F, Karlsson S, Faresjö M, Larsson D. Cis-regulatory elements in conserved non-coding sequences of nuclear receptor genes indicate for crosstalk between endocrine systems. Open Med (Wars) 2021; 16:640-650. [PMID: 33954257 PMCID: PMC8051167 DOI: 10.1515/med-2021-0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors that regulate gene expression when bound to specific DNA sequences. Crosstalk between steroid NR systems has been studied for understanding the development of hormone-driven cancers but not to an extent at a genetic level. This study aimed to investigate crosstalk between steroid NRs in conserved intron and exon sequences, with a focus on steroid NRs involved in prostate cancer etiology. For this purpose, we evaluated conserved intron and exon sequences among all 49 members of the NR Superfamily (NRS) and their relevance as regulatory sequences and NR-binding sequences. Sequence conservation was found to be higher in the first intron (35%), when compared with downstream introns. Seventy-nine percent of the conserved regions in the NRS contained putative transcription factor binding sites (TFBS) and a large fraction of these sequences contained splicing sites (SS). Analysis of transcription factors binding to putative intronic and exonic TFBS revealed that 5 and 16%, respectively, were NRs. The present study suggests crosstalk between steroid NRs, e.g., vitamin D, estrogen, progesterone, and retinoic acid endocrine systems, through cis-regulatory elements in conserved sequences of introns and exons. This investigation gives evidence for crosstalk between steroid hormones and contributes to novel targets for steroid NR regulation.
Collapse
Affiliation(s)
- Maria Araceli Diaz Cruz
- Research School of Health and Welfare, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Dan Lund
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Ferenc Szekeres
- Department of Biomedicine, School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Sandra Karlsson
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Maria Faresjö
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Dennis Larsson
- Sahlgrenska University Hospital, Gothia Forum for Clinical Research, Gothenburg, Sweden
| |
Collapse
|
25
|
Rodríguez-Carrillo A, Rosenmai AK, Mustieles V, Couderq S, Fini JB, Vela-Soria F, Molina-Molina JM, Ferrando-Marco P, Wielsøe M, Long M, Bonefeld-Jorgensen EC, Olea N, Vinggaard AM, Fernández MF. Assessment of chemical mixtures using biomarkers of combined biological activity: A screening study in human placentas. Reprod Toxicol 2021; 100:143-154. [PMID: 33444715 DOI: 10.1016/j.reprotox.2021.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 12/26/2020] [Accepted: 01/04/2021] [Indexed: 11/26/2022]
Abstract
Humans are simultaneously exposed to complex mixtures of chemicals with limited knowledge on potential health effects, therefore improved tools for assessing these mixtures are needed. As part of the Human Biomonitoring for Europe (HBM4EU) Project, we aimed to examine the combined biological activity of chemical mixtures extracted from human placentas using one in vivo and four in vitro bioassays, also known as biomarkers of combined effect. Relevant endocrine activities (proliferative and/or reporter gene assays) and four endpoints were tested: the estrogen receptor (ER), androgen receptor (AR), and aryl hydrocarbon receptor (AhR) activities, as well as thyroid hormone (TH) signaling. Correlations among bioassays and their functional shapes were evaluated. Results showed that all placental extracts agonized or antagonized at least three of the abovementioned endpoints. Most placentas induced ER-mediated transactivation and ER-dependent cell proliferation, together with a strong inhibition of TH signaling and the AR transactivity; while the induction of the AhR was found in only one placental extract. The effects in the two estrogenic bioassays were positively and significantly correlated and the AR-antagonism activity showed a positive borderline-significant correlation with both estrogenic bioassay activities. However, the in vivo anti-thyroid activities of placental extracts were not correlated with any of the tested in vitro assays. Findings highlight the importance of comprehensively mapping the biological effects of "real-world" chemical mixtures present in human samples, through a battery of in vitro and in vivo bioassays. This approach should be a complementary tool for epidemiological studies to further elucidate the combined biological fingerprint triggered by chemical mixtures.
Collapse
Affiliation(s)
- Andrea Rodríguez-Carrillo
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain
| | - Anna Kjerstine Rosenmai
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Vicente Mustieles
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain.
| | - Stephan Couderq
- Physiologie moléculaire et Adaptation, Département "Adaptation du Vivant," UMR 7221 MNHN/CNRS, Muséum National d'Histoire Naturelle, Paris 75005, France
| | - Jean-Baptiste Fini
- Physiologie moléculaire et Adaptation, Département "Adaptation du Vivant," UMR 7221 MNHN/CNRS, Muséum National d'Histoire Naturelle, Paris 75005, France
| | - Fernando Vela-Soria
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain
| | - Jose Manuel Molina-Molina
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain
| | | | - Maria Wielsøe
- Centre for Arctic Health & Molecular Epidemiology, Department of Public Health Aarhus University, Denmark
| | - Manhai Long
- Centre for Arctic Health & Molecular Epidemiology, Department of Public Health Aarhus University, Denmark
| | - Eva Cecilie Bonefeld-Jorgensen
- Centre for Arctic Health & Molecular Epidemiology, Department of Public Health Aarhus University, Denmark; Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland
| | - Nicolás Olea
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain
| | - Anne Marie Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Mariana F Fernández
- University of Granada, Center for Biomedical Research (CIBM), Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016, Granada, Spain; Instituto de Investigación Biosanitaria Ibs GRANADA, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), 18100, Spain.
| |
Collapse
|
26
|
Li H, Song G, Zhou Q, Ran R, Jiang H, Zhang R, Liu Y, Zhang J, Meng L, Ma L, Sun Y, Wang M, Zhou Q, Yan H, Zhou Q, Dong X, Tong Y. Activity of preclinical and phase I clinical trial of a novel androgen receptor antagonist GT0918 in metastatic breast cancer. Breast Cancer Res Treat 2021; 189:725-736. [PMID: 34392453 PMCID: PMC8505310 DOI: 10.1007/s10549-021-06345-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/29/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE To evaluate GT0918, a 2nd-generation AR antagonist, for its AR down-regulation activity among breast cancer patients. METHODS The effect of GT0918 on AR protein expression was evaluated in AR expression breast cancer cells and in breast cancer xenograft model. A 3 + 3 phase I dose-escalation study was launched in Peking University Cancer Hospital. The endpoints included dose finding, safety, pharmacokinetics, and antitumor activity. RESULTS GT0918 was demonstrated to effectively suppress the expression of AR protein and the growth of AR-positive breast cancer tumors in mouse xenograft tumor models. All patients treated with GT0918 were at a QD dose-escalation of five dose levels from 100 to 500 mg. The most common treatment-related AEs of any grade were asthenia, anemia, decreased appetite, increased blood cholesterol, increased blood triglycerides, decreased white blood cell count, and increased low-density lipoprotein. Grade 3 AEs were fatigue (2 of 18, 11.1%), aspartate aminotransferase increase (1 of 18, 5.6%), alanine aminotransferase increase (1 of 18, 5.6%), and neutrophil count decrease (1 of 18, 5.6%). Clinical benefit rate (CBR) in 16 weeks was 23.1% (3/13). Among 7 AR-positive patients, 6 can evaluate efficacy, and 2 completed 23.5- and 25-cycle treatment, respectively (as of 2020/1/20). PK parameters showed a fast absorption profile of GT0918 in the single-dose study. GT0918 and its major metabolite reached steady-state serum concentration levels at day 21 after multiple dosing. CONCLUSION GT0918 can effectively inhibit AR-positive breast cancer tumor growth. GT0918 was demonstrated well tolerated with a favorable PK profile. The suitable dose of GT0918 was 500 mg QD and may provide clinical benefits for AR-positive mBC.
Collapse
Affiliation(s)
- Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142, People's Republic of China.
| | - Guohong Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Qiaoxia Zhou
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Ran Ran
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Hanfang Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Ruyan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Yaxin Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Jiayang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Rd, Haidian district, Beijing, 100142 People’s Republic of China
| | - Luping Meng
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Liandong Ma
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Ye Sun
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Meiyu Wang
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Qingqing Zhou
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Honghua Yan
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Qianxiang Zhou
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Xunwei Dong
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| | - Youzhi Tong
- Kintor Pharmaceutical Limited, No. 20 Songbei Road, Suzhou Industrial Park, Jiangsu, 215123 People’s Republic of China
| |
Collapse
|
27
|
Ahn SK, Jung SY. Current Biomarkers for Precision Medicine in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1187:363-379. [PMID: 33983588 DOI: 10.1007/978-981-32-9620-6_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Breast cancer has become the prototypical solid tumor where targets have been identified within the tumor allowing for personalized approach for systemic therapy. Biomarkers are beginning to play an important role in preparing the way for precision treatment. Mandatory biomarkers for every newly diagnosed case of breast cancer are estrogen receptors and progesterone receptors in selecting patients for endocrine treatment and HER2 for identifying patients likely to benefit from antiHER2 therapy. Although methodological problems exist in the determination of Ki67, because of its clearly established clinical value, wide availability, and low costs relative to the available multianalyte signatures, Ki67 may be used for determining prognosis, especially if values are low or high. Also, the androgen receptor (AR) pathway is emerging as a potential therapeutic target in breast cancer. AR-targeted treatments for breast cancer are in development and have shown promising preliminary results. While, most established biomarkers in breast cancer require tissue samples, serum tumor markers are easily accessible and require a less invasive procedure. Among them, tissue polypeptide-specific antigen (TPS), a specific epitope structure of a peptide in serum associated with human cytokeratin 18, is linked to the proliferative activity of tumors. TPS may be a valuable and independent prognostic biomarker for breast cancer.In order to accelerate progress towards precision treatment for women with breast cancer, we need additional predictive biomarker, especially for enhancing the positive predictive value for endocrine and antiHER2 therapies, as well as biomarkers for predicting response to specific forms of chemotherapy.
Collapse
Affiliation(s)
- Soo Kyung Ahn
- Breast and Thyroid Center, Department of Surgery, Kangnam Sacred Heart Hospital, Hallym University, Seoul, South Korea.
| | - So-Youn Jung
- Center for Breast Cancer, National Cancer Center, Goyang-si, South Korea
| |
Collapse
|
28
|
Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife 2020; 9:59616. [PMID: 33289482 PMCID: PMC7744101 DOI: 10.7554/elife.59616] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17β-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, United States
| | - Molly Nelson Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Alicia R Rothman
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Samim Ali Mondal
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Martin-Paul Agbaga
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Archana Unnikrishnan
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | | | - John Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States
| | - Willard M Freeman
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States.,Oklahoma City Veterans Affairs Medical Center, Oklahoma City, United States
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
29
|
Machek SB, Cardaci TD, Wilburn DT, Willoughby DS. Considerations, possible contraindications, and potential mechanisms for deleterious effect in recreational and athletic use of selective androgen receptor modulators (SARMs) in lieu of anabolic androgenic steroids: A narrative review. Steroids 2020; 164:108753. [PMID: 33148520 DOI: 10.1016/j.steroids.2020.108753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/07/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022]
Abstract
Anabolic androgenic steroids (AAS) are testosterone and testosterone-derivative compounds sporadically employed by athletes and increasingly used recreationally to acquire a competitive edge or improve body composition. Nevertheless, users are subject to undesired side effects majorly associated with tissue-specific androgen receptor (AR) binding-mediated actions. More recently, selective AR modulators (SARMs) have gained popularity towards delivering androgen-associated anabolic actions with hopes of minimal androgenic effects. While several SARMs are in preclinical and clinical phases intended for demographics subject to hypogonadism, muscle wasting, and osteoporosis, several athletic organizations and drug testing affiliates have realized the increasingly widespread use of SARMs amongst competitors and have subsequently banned their use. Furthermore, recreational users are haphazardly acquiring these compounds from the internet and consuming doses several times greater than empirically reported. Unfortunately, online sources are rife with potential contamination, despite a prevailing public opinion suggesting SARMs are innocuous AAS alternatives. Considering each agent has a broad range of supporting evidence in both human and non-human models, it is important to comprehensively evaluate the current literature on commercially available SARMs to gain better understanding of their efficacy and if they can truly be considered a safer AAS alternative. Therefore, the purpose of this review is to discuss the current evidence regarding AAS and SARM mechanisms of action, demonstrate the efficacy of several prominent SARMs in a variety of scientific trials, and theorize on the wide-ranging contraindications and potential deleterious effects, as well as potential future directions regarding acute and chronic SARM use across a broad range of demographics.
Collapse
Affiliation(s)
- Steven B Machek
- Exercise & Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation. Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Thomas D Cardaci
- Exercise & Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation. Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA; Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Dylan T Wilburn
- Exercise & Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation. Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Darryn S Willoughby
- Mayborn College of Health Sciences, School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA.
| |
Collapse
|
30
|
Zhang W, Deng Y, Chen L, Zhang L, Wang Z, Liu R, Zhou Z, Diao J. Comparing the effect of triadimefon and its metabolite on male and female Xenopus laevis: Obstructed growth and gonad morphology. CHEMOSPHERE 2020; 259:127415. [PMID: 32603964 DOI: 10.1016/j.chemosphere.2020.127415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 06/11/2023]
Abstract
Amphibians are the most endangered class of vertebrates. In this study, Xenopus laevis frogs were exposed to 0, 1 and 10 mg/L of triadimefon or triadimenol. After 14 or 28 days of exposure, high levels of triadimefon or triadimenol obstructed the growth of frogs. However, low levels of triadimefon induced the growth of females after the longer period of exposure. We also found that the antioxidant enzyme activity and LDH levels in males were higher than those in females after 28-days exposure. In histopathology tests, triadimenol exerted more effect on the ovary while triadimefon exerted more effect on the testes. Additionally, the levels of Estradiol in all 14-day treatments, except 1 mg/L triadimenol, were significantly decreased, however, there was no difference in testosterone levels. Furthermore, triadimefon and triadimenol disrupted the expression of genes controlling hormone homeostasis and reproduction, and this effect depended on the exposure time and the gender of the organism. Our experiments explored the effect of triadimefon and its metabolite on the gonads of frogs and highlighted the role that pesticides are likely to play in the global decline of amphibians.
Collapse
Affiliation(s)
- Wenjun Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Yuanmingyuan West Road 2, Beijing, 100193, China; Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Yue Deng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Yuanmingyuan West Road 2, Beijing, 100193, China; Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Li Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Yuanmingyuan West Road 2, Beijing, 100193, China; Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Luyao Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Yuanmingyuan West Road 2, Beijing, 100193, China; Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Zikang Wang
- Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Rui Liu
- Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Yuanmingyuan West Road 2, Beijing, 100193, China; Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China
| | - Jinling Diao
- Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing, 100193, China.
| |
Collapse
|
31
|
Rajković-Molek K, Mustać E, Avirović M, Georgev P, Demaria M, Aničić J, Ban J, Babarović E. The expression of calpain-1 and androgen receptor in breast cancer and their correlation with clinicopathological characteristics: An immunohistochemical retrospective study. Pathol Res Pract 2020; 216:153068. [PMID: 32825941 DOI: 10.1016/j.prp.2020.153068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 11/24/2022]
Abstract
Breast cancer is a heterogeneous disease with different biological outcome and ability to acquire resistance to therapy. The calpain family of proteases and androgen receptor (AR) are implicated in breast cancer pathogenesis and progression and are potential targets for novel treatment regimens. The aim of this study was to investigate the expression of calpain-1 and AR in breast cancer and to correlate their expression with clinicopathological variables and prognosis of patients. In this study we enrolled 219 breast cancer patients with long term follow-up information available. Immunohistochemical methods on a tissue microarray were used to investigate expression of calpain-1 and AR in tumor cells. The expression of calpain-1 and AR both differed significantly between the tumor subtypes of patients (p = 0.002 and p = 0.042 respectively). High calpain-1 expression was associated with patient's age over 50 years (p = 0.005) and positive ER status (p = 0.009), but not with other clinicopathological variables. Women with AR negative breast cancers were more likely to be older (p = 0.016), to have bigger tumors (p = 0.032), higher stage of the disease (p = 0.026), presence of exulceration (p = 0.017), negative ER status (p = 0.007) and higher Ki-67 proliferative index (p = 0.027). Calpain-1 expression was not associated with breast cancer specific overall survival in the total cohort of patients, however low calpain-1 expression was associated with adverse survival (p = 0.018) in triple negative subgroup of patients. Low calpain-1 expression was also associated with significantly shorter 5-year disease-free survival in total cohort of patients (p = 0.03). AR status was not associated with overall and disease-free survival of patients. This study has demonstrated that the expression of calpain-1 and androgen receptors are associated with important clinicopathological variables. The expression of calpain-1 was associated with improved disease-free survival of all analyzed patients and with improved overall survival of triple negative breast cancer patients.
Collapse
Affiliation(s)
- Koraljka Rajković-Molek
- Clinical Department of Pathology and Cytology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia.
| | - Elvira Mustać
- Clinical Department of Pathology and Cytology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia; Department of Pathology, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Manuela Avirović
- Clinical Department of Pathology and Cytology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia; Department of Pathology, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Paula Georgev
- Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Martina Demaria
- Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Josip Aničić
- Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Josipa Ban
- Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | - Emina Babarović
- Department of Pathology, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
32
|
Zhang M, Meng X, Guo C, Liu J, Xing Z, Wang X, Wang X. Clinicopathological Relevance and Prognostic Value of Androgen Receptor in Mammary Paget's Disease with Underlying Invasive Ductal Carcinoma. Oncol Res Treat 2020; 43:346-353. [PMID: 32645702 DOI: 10.1159/000507893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 04/15/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Mammary Paget's disease (MPD) is a rare cutaneous manifestation. Epidemiologically, more than half of the MPD patients concurrently have underlying invasive ductal carcinoma (MPD-IDC), and their prognosis remains poor despite multimodal treatments of breast cancer have markedly improved patients' survival. Accordingly, it is crucial to seek out novel therapeutic targets of MPD-IDC. As an emerging biological marker, the value of androgen receptor (AR) in MPD-IDC is inconsistent. Our objectives were to investigate the associations between AR and clinicopathological factors, and to explore its prognostic value in MPD-IDC. METHODS We retrospectively analyzed data from 103 MPD-IDC patients, and immunohistochemical staining was used to determine their AR statuses. RESULTS AR was expressed in 44 patients (42.7%), and AR expression was significantly correlated with body mass index (BMI) (p = 0.038) and axillary lymph node (ALN) status (p = 0.025). Kaplan-Meier curves showed that AR positivity was significantly associated with better overall survival (OS) in MPD-IDC patients (p = 0.019) and estrogen receptor-negative MPD-IDC patients (p = 0.039). Multivariate Cox regression analysis revealed that AR was not an independent prognostic indicator of disease-free survival (DFS) or OS in MPD-IDC patients (p = 0.395 and p = 0.073, respectively). CONCLUSIONS In contrast to AR-negative tumors, patients with AR-positive ones were more likely to have lower BMI, no ALN metastasis, and better OS. AR-targeted treatments for MPD-IDC may add to existing therapeutic approaches to improve their effectiveness.
Collapse
Affiliation(s)
- Menglu Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangzhi Meng
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changyuan Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeyu Xing
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
De Bosscher K, Desmet SJ, Clarisse D, Estébanez-Perpiña E, Brunsveld L. Nuclear receptor crosstalk - defining the mechanisms for therapeutic innovation. Nat Rev Endocrinol 2020; 16:363-377. [PMID: 32303708 DOI: 10.1038/s41574-020-0349-5] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Nuclear receptor crosstalk can be defined as the interplay between different nuclear receptors or between their overlapping signalling pathways. A subset of nuclear receptors (such as PPARs and RARs) engage in the formation of well-characterized 'typical' heterodimers with RXR. 'Atypical' heterodimers (such as GR with PPARs, or PPAR with ERR) might form a novel class of physical complexes that might be more transient in nature. These heterodimers might harbour strong transcriptional flexibility, with no strict need for DNA binding of both partners. Direct crosstalk could stem from a pairwise physical association between atypical nuclear receptor heterodimers, either via pre-existing interaction pairs or via interactions that are newly induced with small molecules; such crosstalk might constitute an uncharted space to target nuclear receptor physiological and/or pathophysiological actions. In this Review, we discuss the emerging aspects of crosstalk in the nuclear receptor field and present various mechanistic crosstalk modes with examples that support applicability of the atypical heterodimer concept. Stabilization or disruption, in a context-dependent or cell type-dependent manner, of these more transient heterodimers is expected to fuel unprecedented translational approaches to yield novel therapeutic agents to treat major human diseases with higher precision.
Collapse
Affiliation(s)
- Karolien De Bosscher
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium.
| | - Sofie J Desmet
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium
| | - Eva Estébanez-Perpiña
- Laboratory of Structural Biology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB) of the University of Barcelona (UB), Barcelona, Spain
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Eindhoven, Netherlands
| |
Collapse
|
34
|
Are Androgen and Estrogen Receptors in DCIS Patients Prognostic Indicators of Relapse Independently of Treatment? Appl Immunohistochem Mol Morphol 2020; 27:301-305. [PMID: 28968271 DOI: 10.1097/pai.0000000000000582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ductal carcinoma in situ (DCIS) is a highly heterogenous tumor that is now more frequently diagnosed because of the increased number of screening programs. Women with DCIS are mainly treated with conservative surgery almost always followed by radiotherapy. Although conventional biomarkers, i.e. ER, PgR, Ki67, and HER2, have been extensively investigated in invasive tumors, little is known about their role in DCIS, especially that of the androgen receptor (AR). In the present study, the expression of conventional biomarkers and AR was determined by immunohistochemistry in 85 DCIS samples from patients monitored for a maximum of 13 years: 43 patients were treated with quadrantectomy and 42 patients underwent quadrantectomy and radiotherapy. Of these, 5 and 11 patients relapsed, respectively. Our findings showed that ER and PgR were higher in nonrelapsed than in relapsed patients (P=0.025 and 0.0038). In contrast, AR expression and the AR/ER ratio were higher in relapsed patients than in the nonrelapsed group (P=0.0069 and 0.0012). At the best cut-off value of 1.1, the AR/ER ratio showed an overall accuracy of 92% and 80% in predicting in situ relapse or progression to invasive carcinoma in DCIS patients treated with surgery and those treated with surgery plus radiotherapy, respectively. AR would therefore appear to be an independent prognostic biomarker in the latter group. Our preliminary results highlight the potentially important role of the AR/ER ratio as a predictive indicator of DCIS relapse, independently of treatment.
Collapse
|
35
|
Fu WF, Li JJ, Kang SH, Song CG. The Expression, Clinicopathologic Characteristics, and Prognostic Value of Androgen Receptor in Breast Cancer: A Bioinformatics Analysis Using Public Databases. DNA Cell Biol 2020; 39:864-874. [PMID: 32181676 DOI: 10.1089/dna.2019.5192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The role of androgen receptor (AR) in breast cancer has been unveiled in succession for the past few years. In this study, we conducted a comprehensive analysis based on four online public databases of data from many previous studies. We found that the expression of AR is significantly related to age, histological grade, and subtype but not to lymph node status. The low expression level of AR is strongly associated with poor recurrence-free survival, especially with poor distance metastasis-free survival in luminal A patients, but inverse in HER2 (human epidermal growth factor receptor-2) enriched patients. AR might be a biomarker of chemosensitivity in the basal subtype. Besides, the expression of melanophilin (MLPH) is distinctly in accordance with that of AR. AR could play diverse roles in different subtypes of breast cancer.
Collapse
Affiliation(s)
- Wen-Fen Fu
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Hong Kang
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| | - Chuan-Gui Song
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
36
|
Amaral C, Augusto TV, Almada M, Cunha SC, Correia-da-Silva G, Teixeira N. The potential clinical benefit of targeting androgen receptor (AR) in estrogen-receptor positive breast cancer cells treated with Exemestane. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165661. [PMID: 31891807 DOI: 10.1016/j.bbadis.2019.165661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 12/09/2019] [Accepted: 12/24/2019] [Indexed: 01/19/2023]
Abstract
The development of acquired resistance to the aromatase inhibitors (AIs) used in clinic is being considered the major concern in estrogen-receptor positive (ER+) breast cancer therapy. Recently, androgen receptor (AR) has gained attention in the clinical setting, since it has been implicated in AIs-resistance, although, different roles for AR in cell fate have been described. In this work, our group elucidates, for the first time, the oncogenic role of AR in sensitive and resistant ER+ breast cancer cells treated with the potent third-generation steroidal AI Exemestane (Exe). We demonstrate that Exe promotes an overexpression/activation of AR, which has an oncogenic and pro-survival role in Exe-sensitive and Exe-resistant cells. Moreover, we also disclose that targeting AR with bicalutamide (CDX) in Exe-treated cells, enhances the efficacy of this AI in sensitive cells and re-sensitizes resistant cells to Exe treatment. Furthermore, by targeting AR in Exe-resistant cells, it is also possible to block the activation of the ERK1/2 and PI3K cell survival pathways, hamper ERα activation and increase ERβ expression. Thus, this study, highlights a new mechanism involved in Exe-acquired resistance, implicating AR as a key molecule in this setting and suggesting that Exe-resistant cells may have an AR-dependent but ER-independent mechanism. Hence we propose AR antagonism as a potential and attractive therapeutic strategy to overcome Exe-acquired resistance or to enhance the growth inhibitory properties of Exe on ER+ breast cancer cells, improving breast cancer treatment.
Collapse
Affiliation(s)
- Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Tiago V Augusto
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Marta Almada
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Sara C Cunha
- LAQV.REQUIMTE, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| |
Collapse
|
37
|
Augusto TV, Amaral C, Varela CL, Bernardo F, da Silva ET, Roleira FFM, Costa S, Teixeira N, Correia-da-Silva G. Effects of new C6-substituted steroidal aromatase inhibitors in hormone-sensitive breast cancer cells: Cell death mechanisms and modulation of estrogen and androgen receptors. J Steroid Biochem Mol Biol 2019; 195:105486. [PMID: 31557516 DOI: 10.1016/j.jsbmb.2019.105486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/05/2019] [Accepted: 09/21/2019] [Indexed: 01/14/2023]
Abstract
Estrogen receptor-positive (ER+) breast cancers require estrogens for their growth. Aromatase inhibitors (AIs) are considered the first-line therapy for this type of tumours. Despite the well-established clinical benefit of this therapy, the search for novel potent AIs that present higher efficacy and fewer side effects is still demanded. Thus, taking into account the known interactions of the natural substrate, androstenedione, within the aromatase active-site, a range of new steroidal compounds have been designed, synthesized and studied by our group. In this work, it was evaluated in MCF-7aro, an ER+ breast cancer cell line that overexpress aromatase, the anti-aromatase efficacy and the biological effects of eight new AIs: 6α-methyl-5α-androst-3-en-17-one (1a), 6α-methyl-3α,4α-epoxy-5α-androstan-17-one (3a), 6α-methylandrost-4-ene-3,17-dione (9), 6α-allylandrosta-1,4-diene-3,17-dione (13), 6α-allylandrost-4-ene-3,17-dione (15), 6α-allylandrost-4-en-17-one (17), 6β-hydroxyandrost-4-ene-3,17-dione (19) and 6α-hydroxyandrost-4-ene-3,17-dione (20). Their anti-cancer properties were elucidated, as well as, the dependence of their mechanism of action on aromatase inhibition and/or on steroid receptors modulation, such as estrogen and androgen receptors, which are key targets for this type of cancer. Results demonstrate that the studied AIs present high anti-aromatase activity, disrupt MCF-7aro cell cycle progression and induce apoptosis, through the mitochondrial pathway. Compounds 1a, 3a, 9, 13, 15 and 17 exhibited an aromatase-dependent effect on cells and, interestingly, steroids 9 and 13 displayed the ability to decrease aromatase protein levels without affecting CYP19A1 mRNA levels. Furthermore, the effects of compounds 1a, 3a and 15 were dependent on ER and on AR modulation, whereas compounds 9 and 19 were only dependent on AR modulation. From a clinical point of view, these actions can be considered as a therapeutic advantage for this type of tumours. Thus, new promising AIs that impair ER+ breast cancer cell growth, by acting on aromatase, and even, on ER and AR were discovered. Furthermore, new insights on the most favourable structural modifications in the steroidal core structure were provided, helping to a more rational drug design of new and potent AIs.
Collapse
Affiliation(s)
- Tiago V Augusto
- Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina Amaral
- Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carla L Varela
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Fernanda Bernardo
- Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Elisiário Tavares da Silva
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Fernanda F M Roleira
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Saul Costa
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Natércia Teixeira
- Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Georgina Correia-da-Silva
- Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
38
|
A high AR:ERα or PDEF:ERα ratio predicts a sub-optimal response to tamoxifen therapy in ERα-positive breast cancer. Cancer Chemother Pharmacol 2019; 84:609-620. [PMID: 31297554 DOI: 10.1007/s00280-019-03891-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 06/13/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Approximately 30% oestrogen receptor alpha (ERα)-positive breast cancer (BC) patients exhibit intrinsic or recurrent resistance to adjuvant endocrine therapy with tamoxifen. The androgen receptor (AR) is expressed in about 90% of ERα-positive patients, with particularly high expression in tamoxifen-resistant tumours. Prostate-derived Ets factor (PDEF), which is a co-regulator of AR, plays a role in tamoxifen resistance in ERα-positive BC. The purpose of this research was to analyse the potential roles of AR, PDEF and ERα levels in the response to tamoxifen resistance in ERα-positive BC. METHODS The nuclear AR:ERα and PDEF:ERα ratios were examined immunohistochemically in a cohort of 225 ERα-positive pre-menopausal BC patients who had received adjuvant tamoxifen therapy. RESULTS For both AR:ERα and PDEF:ERα ratios, the optimal cutoff value was 2.0. Patients receiving adjuvant tamoxifen treatment who had a high AR:ERα (≥ 2.0) (HR = 3.90) or PDEF:ERα ratio (≥ 2.0) (HR = 2.77) had a beyond twofold increased risk of failure. Both the AR:ERα ratio (P = 0.001) and PDEF:ERα ratio (P = 0.002) were independently associated with the risk of tamoxifen treatment failure. Furthermore, both a high ratio of AR:ERα (≥ 2.0) and PDEF:ERα (≥ 2.0) were associated with shorter disease-free survival (DFS) and shorter disease-specific survival (DSS). In addition, both the AR:ERα ratio and PDEF:ERα ratio were independent predictors of DFS (both P < 0.0001) and DSS (P = 0.001 and P < 0.0001, respectively). CONCLUSIONS AR:ERα and PDEF:ERα ratios are independent predictors of the response to conventional ERα-directed tamoxifen endocrine therapy.
Collapse
|
39
|
Estrogen Receptor Positive Breast Cancer with High Expression of Androgen Receptor has Less Cytolytic Activity and Worse Response to Neoadjuvant Chemotherapy but Better Survival. Int J Mol Sci 2019; 20:ijms20112655. [PMID: 31151151 PMCID: PMC6600230 DOI: 10.3390/ijms20112655] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 11/17/2022] Open
Abstract
Estrogen receptor (ER) positive breast cancer (BC), the most abundant BC subtype, is notorious for poor response to neoadjuvant chemotherapy (NAC). The androgen receptor (AR) was reported to support estradiol-mediated ER activity in an in vitro system. Recently, ER-positive BC with fewer tumor infiltrating lymphocytes (TILs) was shown to have a better prognosis, opposite to the trend seen with ER-negative BC. We hypothesized that ER-positive BC with high expression of AR will have fewer TILs and an inferior response to NAC, but with a better prognosis. In both TCGA and METABRIC cohorts, AR expression was significantly higher in ER-positive BCs compared to ER-negatives (p < 0.001, p < 0.001, respectively) and it correlated with ER expression (R = 0.630, R = 0.509, respectively). In ER-positive tumors, AR high tumors enriched UV response down (NES = 2.01, p < 0.001), and AR low tumors enriched DNA repair (NES = −2.02, p < 0.001). AR high tumors were significantly associated with procancer regulatory T-cells, and AR low tumors were associated with anticancer immune cells, such as CD4, CD8, and Gamma-Delta T-cells and memory B-cells in ER-positive BC (p < 0.01). Further, cytolytic activity was significantly lower in AR high BC in both cohorts. Finally, AR high tumors had a significantly lower rate of attaining pathological complete response to NAC (GSE22358), but better survival. In conclusion, our results demonstrated that high AR has fewer tumor infiltrating lymphocytes as well as cytolytic activity and an inferior response to NAC, but better survival in ER-positive BC.
Collapse
|
40
|
Baig MS, Kolasa-Wołosiuk A, Pilutin A, Safranow K, Baranowska-Bosiacka I, Kabat-Koperska J, Wiszniewska B. Finasteride-Induced Inhibition of 5α-Reductase Type 2 Could Lead to Kidney Damage-Animal, Experimental Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16101726. [PMID: 31100850 PMCID: PMC6572442 DOI: 10.3390/ijerph16101726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/25/2022]
Abstract
In the pharmacological treatment of prostate cancer, benign prostatic hyperplasia and androgenetic alopecia finasteride is commonly used. This drug inhibits 5α-reductase type 2, which is why finasteride affects androgen homeostasis, since testosterone (T) cannot be reduced to dihydrotestosterone (DHT). As studies on sex-related renal injuries suggest a high probability of androgen-induced renal dysfunction, the aim of this study was to determine the potential harmful effects of finasteride on the kidneys of rats. The study was performed on sexually mature male Wistar rats given finasteride. Histological sections of the kidneys were used for immunohistochemical visualization of the androgen receptor (AR), junctional proteins (occluding (Occ); E-cad, N-cad, E-/N-cadherin; β-cat, β-catenin; connexin 43 (Cx43)), proliferating cell nuclear antigen (PCNA), IL-6, and lymphocyte markers (CD3 for T cell, CD19 for B cell). The TUNEL method was used for cell apoptosis identification, and picro sirius red staining was used to assess collagen fibers thickness. The levels of T, DHT and estradiol (E2) were determined in blood serum. It was shown that finasteride treatment affected steroid hormone homeostasis, altered the expression of AR and intracellular junction proteins, changed the ratio between cell apoptosis and proliferation, and caused lymphocyte infiltration and an increase of IL-6. The thickening of collagen fibers was observed as tubular fibrosis and glomerulosclerosis. Summarizing, finasteride-induced hormonal imbalance impaired the morphology (i.e., dysplastic glomeruli, swollen proximal convoluted tubules) and physiology (changed level of detected proteins/markers expression) of the kidneys. Therefore, it is suggested that patients with renal dysfunction or following renal transplantation, with androgen or antiandrogen supplementation, should be under special control and covered by extended diagnostics, because the adverse negative effect of DHT deficiency on the progression of kidney disease cannot be ignored.
Collapse
Affiliation(s)
- Mirza Saim Baig
- Department of Histology and Embryology, Pomeranian Medical University, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Agnieszka Kolasa-Wołosiuk
- Department of Histology and Embryology, Pomeranian Medical University, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Anna Pilutin
- Department of Histology and Embryology, Pomeranian Medical University, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Joanna Kabat-Koperska
- Department of Nephrology, Transplantology and Internal Medicine Pomeranian Medical University, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University, Powst. Wlkp. 72, 70-111 Szczecin, Poland.
| |
Collapse
|
41
|
Anestis A, Sarantis P, Theocharis S, Zoi I, Tryfonopoulos D, Korogiannos A, Koumarianou A, Xingi E, Thomaidou D, Kontos M, Papavassiliou AG, Karamouzis MV. Estrogen receptor beta increases sensitivity to enzalutamide in androgen receptor-positive triple-negative breast cancer. J Cancer Res Clin Oncol 2019; 145:1221-1233. [PMID: 30805773 DOI: 10.1007/s00432-019-02872-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/20/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Androgen receptor (AR) is playing an important role in the progression of a subset of TNBC. We evaluated the impact of ERβ expression along with anti-AR drugs in AR-positive TNBC. METHODS ERβ expression was examined in AR-positive TNBC cell line using MTT assay, scratch and Annexin V-FITC assay in the presence or absence of anti-androgens. Protein levels of involved molecules were assessed using Western blot. Receptors' localization was detected by immunofluorescence and their physical association was examined using proximity ligation assay (PLA), which enables the visualization of interacting proteins in fixed cells and tissues. RESULTS Transient transfection of ERβ in MDA-MB 453 AR-positive TNBC cell line significantly inhibited cell proliferation, metastatic potential and induced apoptosis. ERβ expression reversed the aggravating role of AR in both indirect and direct ways. Indirectly, ERβ decreased AR activation through the inhibition of PI3K/AKT signaling pathway. Directly, ERβ formed heterodimers with AR in MDA-MB 453 cells and in human tissue samples impeding AR from forming homodimers. Enzalutamide is a more potent anti-androgen in AR + TNBC compared to bicalutamide. ERβ expression increased the sensitivity of MDA-MB 453 cells to anti-androgens and especially to enzalutamide. The administration of enzalutamide enhanced AR:ERβ heterodimers formation increasing the anti-tumor capacity of ERβ. CONCLUSIONS Collectively, our results provide evidence for a novel mechanism by which ERβ exerts oncosuppressive effect in AR-positive TBNC through direct and indirect interactions with AR. Moreover, ERβ expression may identify a new subset of TNBC that would respond more favorable to anti-androgens.
Collapse
Affiliation(s)
- Aristomenis Anestis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Stamatios Theocharis
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Ilianna Zoi
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | | | | | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Xingi
- Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece
| | | | - Michalis Kontos
- First Department of Surgery, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece.
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece.
- First Department of Internal Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
42
|
Abstract
Male breast cancer is rare, accounting for 1% of all breast cancer diagnoses in the USA. Because of its rarity, most major breast cancer trials have included only female patients. This has resulted in limited prospective data to guide the clinical management of men with breast cancer. As a result, treatment decisions are typically extrapolated from data generated in female patients. This approach may be suboptimal, particularly considering the differing hormonal milieus between men and women with respect to both breast cancer development and treatment. Herein, we summarize current knowledge of the biology and clinicopathology of male breast cancer and review current approaches to locoregional and systemic management of this rare disease.
Collapse
|
43
|
Vasiliou SK, Diamandis EP. Androgen receptor: A promising therapeutic target in breast cancer. Crit Rev Clin Lab Sci 2019; 56:200-223. [PMID: 30821186 DOI: 10.1080/10408363.2019.1575643] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Breast cancer (BCa) is the second most common cancer worldwide and the most prevalent cancer in women. The majority of BCa cases are positive (+) for the estrogen receptor (ER+, 80%) and progesterone receptor (PR+, 65%). Estrogen and progesterone hormones are known to be involved in cancer progression, and thus hormonal deprivation is used as an effective treatment for ER+PR+ BCa subtypes. However, some ER+PR+ BCa patients develop resistance to such therapies. Meanwhile, chemotherapy is the only available treatment for ER-PR- BCa tumors. Another hormone receptor known as the androgen receptor (AR) has also been found to be widely expressed in human breast carcinomas. However, the mechanisms of AR and its endogenous androgen ligands is not well-understood in BCa and its biological role in this hormone-related disease remains unclear. In this review, we aim to address the importance of the AR in BCa diagnosis and prognosis, current AR-targeting approaches in BCa, and the potential for AR-downstream molecules to serve as therapeutic targets.
Collapse
Affiliation(s)
- Stella K Vasiliou
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada
| |
Collapse
|
44
|
Ji W, Shi Y, Wang X, He W, Tang L, Tian S, Jiang H, Shu Y, Guan X. Combined Androgen receptor blockade overcomes the resistance of breast cancer cells to palbociclib. Int J Biol Sci 2019; 15:522-532. [PMID: 30745839 PMCID: PMC6367574 DOI: 10.7150/ijbs.30572] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/28/2018] [Indexed: 02/03/2023] Open
Abstract
The dysregulation of cyclin D -Cyclin-dependent kinase 4/6 (CDK4/6)-Rb axis has been implicated in breast cancer progression and the selective CDK4/6 inhibitors have shown effective activity in advanced breast cancer, especially in tumors driven by the estrogen receptor (ER). However, resistance to these small molecular inhibitors has become an inevitable clinical issue after their initial use. Here, we investigated the potential mechanism of resistance by establishing a CDK4/6 inhibitor palbociclib-resistant breast cancer cell line (MCF-7pR). After prolonged exposure to palbociclib, we detected the loss of the ER signaling and an increase in androgen receptor (AR). Moreover, we demonstrated more localization of AR in the cell nucleus of MCF-7pR compared to the parental cell (MCF-7). We also reported that AR could promote the progression of the cell cycle. Blockade of AR signaling could reduce the level of the relative G1-S cyclins, abolish Rb phosphorylation and inhibit the activation of transcriptional programs in S phase. Furthermore, dual inhibition of AR and CDK4/6 could reverse the resistance of palbociclib both in vitro and in vivo. In sum, our studies provide evidence that AR activation promotes cell cycle progression and cell proliferation in CDK4/6 inhibitor resistance, and identify AR inhibition as a putative novel therapeutic strategy to treat CDK4/6 inhibitor resistance in cancer.
Collapse
Affiliation(s)
- Wenfei Ji
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing 210002, China
| | - Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Xin Wang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Weiwei He
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing 210002, China
| | - Lin Tang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Shengwang Tian
- Department of Oncology, JinTan People's Hospital, Jintan 213200, China
| | - Hua Jiang
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital with Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing 210002, China.,Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
45
|
Kuo PL, Tseng JY, Chen HI, Wu CY, Omar HA, Wang CY, Cheng HY, Hsu CC, Fu TF, Teng YN. Identification of SEPTIN12 as a novel target of the androgen and estrogen receptors in human testicular cells. Biochimie 2018; 158:1-9. [PMID: 30513371 DOI: 10.1016/j.biochi.2018.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/29/2018] [Indexed: 11/16/2022]
Abstract
SEPTIN12 (SEPT12) is a testis-enriched gene that is downregulated in the testis of infertile men with severe spermatogenic defects. While SEPT12 is involved in spermatogenic failure and sperm motility disorder, SEPT12 transcriptional regulation is still unknown. Here we report the promoter region of SEPT12 as a 245 bp segment upstream of the transcription start site. One androgen receptor (AR) and two estrogen receptor α (ERα) binding sites in this region were initially identified by bioinformatics prediction and confirmed by chromatin immunoprecipitation assay. Truncated ERα or AR binding sites decreased the promoter activity, which indicated that the ERα and AR are essential for the SEPT12 promoter. On the other hand, the promoter activity was enhanced by the treatment with 17β-estradiol (E2) and 5α-dihydrotestosterone (5α-DHT). Thus, one androgen and two estrogen hormone responsive elements located in the promoter of SEPT12 gene can regulate SEPT12 expression. Two single nucleotide polymorphisms (SNPs), rs759992 T > C and rs3827527 C > T, were observed in the SEPT12 gene promoter region and were able to decrease the promoter activity. In conclusion, the current work identified the promoter of the human SEPT12 gene and provided key evidence about its transcriptional regulation via E2 and 5α-DHT. Since SEPT12 has an important role in spermatogenesis, SEPT12 expression analysis can be developed as a potential tool for the assessment of environmental or food pollution by hormones or for the evaluation of the risk of endocrine-disrupting chemicals (EDCs) in general.
Collapse
Affiliation(s)
- Pao-Lin Kuo
- Department of Obstetrics & Gynecology, National Cheng Kung University, College of Medicine, Tainan, 701, Taiwan
| | - Jie-Yun Tseng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, 700, Taiwan
| | - Hau-Inh Chen
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Chia-Yun Wu
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, 700, Taiwan
| | - Hany A Omar
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, College of Medicine, Tainan, 701, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, 701, Taiwan
| | - Han-Yi Cheng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, 700, Taiwan
| | - Chao-Chin Hsu
- Institute of Reproductive Medicine, Taipei Medical University Hospital, Taipei, 110, Taiwan
| | - Tzu-Fun Fu
- Institute of Basic Medical Sciences, National Cheng Kung University, College of Medicine, Tainan, 701, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, 700, Taiwan.
| |
Collapse
|
46
|
Truong TH, Lange CA. Deciphering Steroid Receptor Crosstalk in Hormone-Driven Cancers. Endocrinology 2018; 159:3897-3907. [PMID: 30307542 PMCID: PMC6236424 DOI: 10.1210/en.2018-00831] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/04/2018] [Indexed: 12/27/2022]
Abstract
Steroid hormone receptors (SRs) have a multitude of functions in human biology and disease progression. The SR family of related ligand-activated transcription factors includes androgen, estrogen, glucocorticoid, mineralocorticoid, and progesterone receptors. Antiestrogen or estrogen receptor (ER)-targeted therapies to block ER action remain the primary treatment of luminal breast cancers. Although this strategy is successful, ∼40% of patients eventually relapse due to endocrine resistance. The majority of hormone-independent tumors retain some level of SR expression, but sidestep hormone ablation treatments. SRs are known to crosstalk extensively with kinase signaling pathways, and this interplay has been shown to bypass ER-targeted therapies in part by providing alternative proliferation and survival signals that enable hormone independence. Modified receptors adopt alternate conformations that resist antagonism or promote agonism. SR-regulated transcription and SR-binding events have been classically studied as single receptor events using single hormones. However, it is becoming increasingly evident that individual steroids and SRs rarely act alone. Emerging evidence shows that coexpressed SRs crosstalk with each other in hormone-driven cancers, such as breast and prostate. Crosstalk between related SRs allows them to modulate signaling and transcriptional responses to noncognate ligands. This flexibility can lead to altered genomic binding and subsequent changes in SR target gene expression. This review will discuss recent mechanistic advances in elucidating SR crosstalk and the implications for treating hormone-driven cancers. Understanding this crosstalk (i.e., both opposing and collaborative) is a critical step toward expanding and modernizing endocrine therapies and will ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Thu H Truong
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
47
|
Bleach R, McIlroy M. The Divergent Function of Androgen Receptor in Breast Cancer; Analysis of Steroid Mediators and Tumor Intracrinology. Front Endocrinol (Lausanne) 2018; 9:594. [PMID: 30416486 PMCID: PMC6213369 DOI: 10.3389/fendo.2018.00594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.
Collapse
Affiliation(s)
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
48
|
Siersbæk R, Kumar S, Carroll JS. Signaling pathways and steroid receptors modulating estrogen receptor α function in breast cancer. Genes Dev 2018; 32:1141-1154. [PMID: 30181360 PMCID: PMC6120708 DOI: 10.1101/gad.316646.118] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Estrogen receptor α (ER) is the major driver of ∼75% of breast cancers, and multiple ER targeting drugs are routinely used clinically to treat patients with ER+ breast cancer. However, many patients relapse on these targeted therapies and ultimately develop metastatic and incurable disease, and understanding the mechanisms leading to drug resistance is consequently of utmost importance. It is now clear that, in addition to estrogens, ER function is modulated by other steroid receptors and multiple signaling pathways (e.g., growth factor and cytokine signaling), and many of these pathways affect drug resistance and patient outcome. Here, we review the mechanisms through which these pathways impact ER function and drug resistance as well as discuss the clinical implications.
Collapse
Affiliation(s)
- Rasmus Siersbæk
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Sanjeev Kumar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
49
|
Takagi K, Miki Y, Ishida T, Sasano H, Suzuki T. The interplay of endocrine therapy, steroid pathways and therapeutic resistance: Importance of androgen in breast carcinoma. Mol Cell Endocrinol 2018; 466:31-37. [PMID: 28918115 DOI: 10.1016/j.mce.2017.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/04/2017] [Accepted: 09/11/2017] [Indexed: 12/27/2022]
Abstract
A great majority of breast carcinomas expresses estrogen receptor (ER) and estrogens have crucial roles in the progress of breast carcinomas. Endocrine therapy targeting ER and/or intratumoral estrogen production significantly improved clinical outcomes of the patients with ER-positive breast carcinomas. However, resistance to endocrine therapy is often observed and significant number of patients will recur after the treatment. In addition, treatment for the patients with triple-negative breast carcinomas (negative for all ER, progesterone receptor (PR) and HER2) are limited to cytotoxic chemotherapy and novel therapeutic targets need to be identified. In breast carcinoma tissues, not only ER but androgen receptor (AR) is frequently expressed, suggesting pivotal roles of androgens in the progress of breast carcinomas. Growing interest on androgen action as possible therapeutic target has been taken, but androgen action seems quite complicated in breast carcinomas and inconsistent findings has been also proposed. In this review, we will summarize recent studies regarding intratumoral androgen production and its regulation as well as distinct subset of breast carcinomas characterized by activated AR signaling and recent clinical trial targeting AR in the patients with either ER-positive and ER-negative breast carcinomas.
Collapse
Affiliation(s)
- Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
50
|
Jia X, Liu Z, Lu X, Tang J, Wu Y, Du Q, He J, Zhang X, Jiang J, Liu W, Zheng Y, Ding Y, Zhu W, Zhang H. Effects of MCLR exposure on sex hormone synthesis and reproduction-related genes expression of testis in male Rana nigromaculata. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 236:12-20. [PMID: 29414332 DOI: 10.1016/j.envpol.2018.01.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 06/08/2023]
Abstract
Microcystin-leucine-arginine (MCLR) is the most popular and toxic variant among microcystins, which can cause severe reproductive toxicity to animals. However, the mechanisms of reproductive toxicity induced by MCLR in amphibians are still not entirely clear. In the current study, toxicity mechanisms of MCLR on the reproductive system of male Rana nigromaculata followed by low concentration (0, 0.1, 1, and 10 μg/L) and short-term (0, 7, and 14 days) MCLR exposure were shown. It was observed that MCLR could be bioaccumulated in the testes of male frogs, and the theoretical bioaccumulation factor values were 0.24 and 0.19 exposed to 1 μg/L and 10 μg/L MCLR for 14 days, respectively. MCLR exposure significantly decreased testosterone (T) concentrations and increased estradiol (E2) concentrations exposed to 1 and 10 μg/L MCLR for 14 days. The mRNA levels of HSD17B3 were downregulated, and HSD17B1 and CYP19A1 mRNA expression levels were upregulated, respectively. Only 10 μg/L MCLR group showed significant induction of follicle-stimulating hormone (FSH) levels and cyclic adenosine monophosphate (cAMP) content. Moreover, AR and ESR1 mRNA expression levels were significantly upregulated exposed to 1 and 10 μg/L MCLR for 14 days, respectively. Our results suggested that low-concentration MCLR induced transcription changes of CYP19A1, HSD17B3, and HSD17B1 led to endocrine disorders, and caused interference of spermatogenesis by the decrease of T and abnormal gene expressions of AR and ESR1 in the testes of R. nigromaculata.
Collapse
Affiliation(s)
- Xiuying Jia
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Zhengquan Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Xiangjun Lu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Juan Tang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Yingzhu Wu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Qiongxia Du
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Jianbo He
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Xinyun Zhang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Jinxiao Jiang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Wenli Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Yuqing Zheng
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Ying Ding
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China
| | - Weiqin Zhu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China
| | - Hangjun Zhang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Guangzhou Key Laboratory of Environmental Exposure and Health, School of Environment, Jinan University, Guangzhou 510632, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China.
| |
Collapse
|